1
|
Gomez-Frittelli J, Devienne GF, Travis L, Kyloh MA, Duan X, Hibberd TJ, Spencer NJ, Huguenard JR, Kaltschmidt JA. Synaptic cell adhesion molecule Cdh6 identifies a class of sensory neurons with novel functions in colonic motility. eLife 2025; 13:RP101043. [PMID: 40193178 PMCID: PMC11975370 DOI: 10.7554/elife.101043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Intrinsic sensory neurons are an essential part of the enteric nervous system (ENS) and play a crucial role in gastrointestinal tract motility and digestion. Neuronal subtypes in the ENS have been distinguished by their electrophysiological properties, morphology, and expression of characteristic markers, notably neurotransmitters and neuropeptides. Here, we investigated synaptic cell adhesion molecules as novel cell-type markers in the ENS. Our work identifies two type II classic cadherins, Cdh6 and Cdh8, specific to sensory neurons in the mouse colon. We show that Cdh6+ neurons demonstrate all other distinguishing classifications of enteric sensory neurons including marker expression of Calcb and Nmu, Dogiel type II morphology and AH-type electrophysiology and IH current. Optogenetic activation of Cdh6+ sensory neurons in distal colon evokes retrograde colonic motor complexes (CMCs), while pharmacologic blockade of rhythmicity-associated current IH disrupts the spontaneous generation of CMCs. These findings provide the first demonstration of selective activation of a single neurochemical and functional class of enteric neurons and demonstrate a functional and critical role for sensory neurons in the generation of CMCs.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford UniversityStanfordUnited States
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
| | - Gabrielle Frederique Devienne
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurology & Neurological Sciences, Stanford UniversityStanfordUnited States
| | - Lee Travis
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Melinda A Kyloh
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San FranciscoSan FranciscoUnited States
| | - Tim J Hibberd
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders Health & Medical Research Institute, Flinders UniversityAdelaideAustralia
| | - John R Huguenard
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurology & Neurological Sciences, Stanford UniversityStanfordUnited States
| | - Julia A Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
- Department of Neurosurgery, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
2
|
Hamnett R, Bendrick JL, Saha Z, Robertson K, Lewis CM, Marciano JH, Zhao ET, Kaltschmidt JA. Enteric glutamatergic interneurons regulate intestinal motility. Neuron 2025; 113:1019-1035.e6. [PMID: 39983724 PMCID: PMC11968238 DOI: 10.1016/j.neuron.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/14/2024] [Accepted: 01/23/2025] [Indexed: 02/23/2025]
Abstract
The enteric nervous system (ENS) controls digestion autonomously via a complex neural network within the gut wall. Enteric neurons expressing glutamate have been identified by transcriptomic studies as a distinct subpopulation, and glutamate can affect intestinal motility by modulating enteric neuron activity. However, the nature of glutamatergic neurons, their position within the ENS circuit, and their function in regulating gut motility are unknown. We identify glutamatergic neurons as longitudinally projecting descending interneurons in the small intestine and colon and as a novel class of circumferential neurons only in the colon. Both populations make synaptic contact with diverse neuronal subtypes and signal with multiple neurotransmitters and neuropeptides in addition to glutamate, including acetylcholine and enkephalin. Knocking out the glutamate transporter VGLUT2 from enkephalin neurons disrupts gastrointestinal transit, while ex vivo optogenetic stimulation of glutamatergic neurons initiates colonic propulsive motility. Our results posit glutamatergic neurons as key interneurons that regulate intestinal motility.
Collapse
Affiliation(s)
- Ryan Hamnett
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| | - Jacqueline L Bendrick
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Zinnia Saha
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Cheyanne M Lewis
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Jack H Marciano
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Stanford Neurosciences Interdepartmental Program, Stanford University, Stanford, CA 94305, USA
| | - Eric Tianjiao Zhao
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
3
|
Hazart D, Moulzir M, Delhomme B, Oheim M, Ricard C. Imaging the enteric nervous system. Front Neuroanat 2025; 19:1532900. [PMID: 40145027 PMCID: PMC11937143 DOI: 10.3389/fnana.2025.1532900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
The enteric nervous system (ENS) has garnered increasing scientific interest due to its pivotal role in digestive processes and its involvement in various gastrointestinal and central nervous system (CNS) disorders, including Crohn's disease, Parkinson's disease, and autism. Despite its significance, the ENS remains relatively underexplored by neurobiologists, primarily because its structure and function are less understood compared to the CNS. This review examines both pioneering methodologies that initially revealed the intricate layered structure of the ENS and recent advancements in studying its three-dimensional (3-D) organization, both in fixed samples and at a functional level, ex-vivo or in-vivo. Traditionally, imaging the ENS relied on histological techniques involving sequential tissue sectioning, staining, and microscopic imaging of single sections. However, this method has limitations representing the full complexity of the ENS's 3-D meshwork, which led to the development of more intact preparations, such as whole-mount preparation, as well as the use of volume imaging techniques. Advancements in 3-D imaging, particularly methods like spinning-disk confocal, 2-photon, and light-sheet microscopies, combined with tissue-clearing techniques, have revolutionized our understanding of the ENS's fine structure. These approaches offer detailed views of its cellular architecture, including interactions among various cell types, blood vessels, and lymphatic vessels. They have also enhanced our comprehension of ENS-related pathologies, such as inflammatory bowel disease, Hirschsprung's disease (HSCR), and the ENS's involvement in neurodegenerative disorders like Parkinson's (PD) and Alzheimer's diseases (AD). More recently, 2-photon or confocal in-vivo imaging, combined with transgenic approaches for calcium imaging, or confocal laser endomicroscopy, have opened new avenues for functional studies of the ENS. These methods enable real-time observation of enteric neuronal and glial activity and their interactions. While routinely used in CNS studies, their application to understanding local circuits and signals in the ENS is relatively recent and presents unique challenges, such as accommodating peristaltic movements. Advancements in 3-D in-vivo functional imaging are expected to significantly deepen our understanding of the ENS and its roles in gastrointestinal and neurological diseases, potentially leading to improved diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Doriane Hazart
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
- Doctoral School Brain, Cognition and Behaviour – ED3C - ED 158, Paris, France
| | - Marwa Moulzir
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
| | - Brigitte Delhomme
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
| | - Martin Oheim
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
| | - Clément Ricard
- Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
| |
Collapse
|
4
|
Ko EA, Zhou T, Ko JH, Jung SC. Transcriptomic Alteration in the Brain and Gut of Offspring Following Prenatal Exposure to Corticosterone. Exp Neurobiol 2025; 34:9-19. [PMID: 40091635 PMCID: PMC11919639 DOI: 10.5607/en24029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Maternal stress during pregnancy can profoundly affect offspring health, increasing the risk of psychiatric disorders, metabolic diseases, and gastrointestinal problems. In this study, the effects of high prenatal corticosterone exposure on gene expression in the brain and small intestine of rat offspring were investigated via RNA-sequencing analysis. Pregnant rats were divided into two groups: Corti.Moms were injected with corticosterone daily, while Nor.Moms were given saline injections. Their offspring were labeled as Corti.Pups and Nor.Pups, respectively. The brain tissue analysis of Corti.Pups showed that the expression levels of the genes linked to neurodegenerative conditions increased and enhanced mitochondrial biogenesis, possibly due to higher ATP demands. The genes associated with calcium signaling pathways, neuroactive ligand-receptor interactions, and IgA production were also upregulated in the small intestine of Corti.pups. Conversely, the genes related to protein digestion, absorption, and serotonergic and dopaminergic synaptic activities were downregulated. These findings revealed that gene expression patterns in both the brain and intestinal smooth muscle of offspring prenatally exposed to corticosterone were substantially altered. Thus, this study provided valuable insights into the effects of prenatal stress on neurodevelopment and gut function.
Collapse
Affiliation(s)
- Eun-A Ko
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Sung-Cherl Jung
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
5
|
Zhang H, Tian Y, Xu C, Chen M, Xiang Z, Gu L, Xue H, Xu Q. Crosstalk between gut microbiotas and fatty acid metabolism in colorectal cancer. Cell Death Discov 2025; 11:78. [PMID: 40011436 DOI: 10.1038/s41420-025-02364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy globally and the second leading cause of cancer-related mortality. Its development is a multifactorial and multistage process influenced by a dynamic interplay between gut microbiota, environmental factors, and fatty acid metabolism. Dysbiosis of intestinal microbiota and abnormalities in microbiota-associated metabolites have been implicated in colorectal carcinogenesis, highlighting the pivotal role of microbial and metabolic interactions. Fatty acid metabolism serves as a critical nexus linking dietary patterns with gut microbial activity, significantly impacting intestinal health. In CRC patients, reduced levels of short-chain fatty acids (SCFAs) and SCFA-producing bacteria have been consistently observed. Supplementation with SCFA-producing probiotics has demonstrated tumor-suppressive effects, while therapeutic strategies aimed at modulating SCFA levels have shown potential in enhancing the efficacy of radiation therapy and immunotherapy in both preclinical and clinical settings. This review explores the intricate relationship between gut microbiota, fatty acid metabolism, and CRC, offering insights into the underlying mechanisms and their potential translational applications. Understanding this interplay could pave the way for novel diagnostic, therapeutic, and preventive strategies in the management of CRC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Yuan Tian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Chunjie Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Miaomiao Chen
- Department of Radiology, Huashan Hospital, National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200040, PR China
| | - Zeyu Xiang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Hanbing Xue
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Qing Xu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
6
|
Yuan X, Ouedraogo SY, Jammeh ML, Simbiliyabo L, Jabang JN, Jaw M, Darboe A, Tan Y, Bajinka O. Can microbiota gut-brain axis reverse neurodegenerative disorders in human? Ageing Res Rev 2025; 104:102664. [PMID: 39818235 DOI: 10.1016/j.arr.2025.102664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
The trillions of microbial populations residing in the gut have recently shown that they can be used as a remedy for various diseases. The gut microbiota-brain-axis interface is one unique pathway that the microbiota demonstrates its medicinal value. This medicinal value is further seen when there is a decline in gut microbial diversity (dysbiosis). Dysbiosis leads to neurodegenerative disorders (NDDs). The objective of this review is to ascertain the clinical significance of gut microbiota induced therapeutic strategies. While navigating this important area of interest, we will elucidate the research gaps, the prospects and the potential reverse interventions of the studied NDDs. In addition to our previous work, relevant literature published in English were searched and retrieved from the PubMed database. The 'gut microbiota and Neurodegenerative disorders' were used as keywords during the search period. The Filters applied are: Abstract, Full text, Meta-Analysis, Randomized Controlled Trial, Reviews, in the last 5 years. The articles were analyzed in our unrelenting quest to make sense of the prospects and research gap in gut microbiota-brain-axis. This chapter is a result of this meticulous work. More convincing data from researches on gut microbiota-brain-axis are required to provide clinical significance including neuroimaging studies. Addressing the structural (pathological footprints) and the functional changes (diseases manifestation) involving gut microbiota-brain-axis require a holistic approach. While the pharmacological therapies such as chemotherapeutic and chemobiotic treatment approaches come with low success rates, non-pharmacological interventions are found to be more useful in reversing NDDs. The inability to detect NDDs at an early stage in their clinical history, makes preventive medicinal approaches the must needed and best intervention strategy. Gut-driven treatments have a lot to offer in the management of refractory neurologic diseases.
Collapse
Affiliation(s)
- Xingxing Yuan
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin 150040, China; Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin 150006, China
| | - Serge Yannick Ouedraogo
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117, China
| | - Modou Lamin Jammeh
- School of Medicine and Allied Health Sciences, University of The Gambia, Gambia
| | - Lucette Simbiliyabo
- Department of Medical Microbiology, Central South University, Changsha, Hunan Provinces, China
| | - John Nute Jabang
- School of Medicine and Allied Health Sciences, University of The Gambia, Gambia
| | - Mariam Jaw
- School of Medicine and Allied Health Sciences, University of The Gambia, Gambia
| | - Alansana Darboe
- Vaccine & Immunity Theme, Infant Immunology, Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine (MRCG@LSHTM), Gambia
| | - Yurong Tan
- Department of Medical Microbiology, Central South University, Changsha, Hunan Provinces, China.
| | - Ousman Bajinka
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117, China; School of Medicine and Allied Health Sciences, University of The Gambia, Gambia; Department of Medical Microbiology, Central South University, Changsha, Hunan Provinces, China.
| |
Collapse
|
7
|
Breßer M, Siemens KD, Schneider L, Lunnebach JE, Leven P, Glowka TR, Oberländer K, De Domenico E, Schultze JL, Schmidt J, Kalff JC, Schneider A, Wehner S, Schneider R. Macrophage-induced enteric neurodegeneration leads to motility impairment during gut inflammation. EMBO Mol Med 2025; 17:301-335. [PMID: 39762650 PMCID: PMC11822118 DOI: 10.1038/s44321-024-00189-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/14/2025] Open
Abstract
Current studies pictured the enteric nervous system and macrophages as modulators of neuroimmune processes in the inflamed gut. Expanding this view, we investigated the impact of enteric neuron-macrophage interactions on postoperative trauma and subsequent motility disturbances, i.e., postoperative ileus. In the early postsurgical phase, we detected strong neuronal activation, followed by transcriptional and translational signatures indicating neuronal death and synaptic damage. Simultaneously, our study revealed neurodegenerative profiles in macrophage-specific transcriptomes after postoperative trauma. Validating the role of resident and monocyte-derived macrophages, we depleted macrophages by CSF-1R-antibodies and used CCR2-/- mice, known for reduced monocyte infiltration, in POI studies. Only CSF-1R-antibody-treated animals showed decreased neuronal death and lessened synaptic decay, emphasizing the significance of resident macrophages. In human gut samples taken early and late during abdominal surgery, we substantiated the mouse model data and found reactive and apoptotic neurons and dysregulation in synaptic genes, indicating a species' overarching mechanism. Our study demonstrates that surgical trauma activates enteric neurons and induces neurodegeneration, mediated by resident macrophages, introducing neuroprotection as an option for faster recovery after surgery.
Collapse
Affiliation(s)
- Mona Breßer
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kevin D Siemens
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Linda Schneider
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | | - Patrick Leven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Tim R Glowka
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristin Oberländer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Elena De Domenico
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE). PRECISE Platform for Genomics and Epigenomics at DZNE and University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim Schmidt
- Department of General, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Jörg C Kalff
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Anja Schneider
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- University of Bonn Medical Center, Dept. of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, 53127, Bonn, Germany
| | - Sven Wehner
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
8
|
Gomez-Frittelli J, Devienne G, Travis L, Kyloh MA, Duan X, Hibberd TJ, Spencer NJ, Huguenard JR, Kaltschmidt JA. Synaptic cell adhesion molecule Cdh6 identifies a class of sensory neurons with novel functions in colonic motility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606748. [PMID: 39149241 PMCID: PMC11326146 DOI: 10.1101/2024.08.06.606748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Intrinsic sensory neurons are an essential part of the enteric nervous system (ENS) and play a crucial role in gastrointestinal tract motility and digestion. Neuronal subtypes in the ENS have been distinguished by their electrophysiological properties, morphology, and expression of characteristic markers, notably neurotransmitters and neuropeptides. Here we investigated synaptic cell adhesion molecules as novel cell type markers in the ENS. Our work identifies two Type II classic cadherins, Cdh6 and Cdh8, specific to sensory neurons in the mouse colon. We show that Cdh6+ neurons demonstrate all other distinguishing classifications of enteric sensory neurons including marker expression of Calcb and Nmu, Dogiel type II morphology and AH-type electrophysiology and I H current. Optogenetic activation of Cdh6+ sensory neurons in distal colon evokes retrograde colonic motor complexes (CMCs), while pharmacologic blockade of rhythmicity-associated current I H disrupts the spontaneous generation of CMCs. These findings provide the first demonstration of selective activation of a single neurochemical and functional class of enteric neurons, and demonstrate a functional and critical role for sensory neurons in the generation of CMCs.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford University; Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
| | - Gabrielle Devienne
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Melinda A. Kyloh
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Xin Duan
- Department of Ophthalmology, School of Medicine, University of California San Francisco; San Francisco, CA, USA
| | - Tim J. Hibberd
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - Nick J. Spencer
- College of Medicine and Public Health, Flinders University; Adelaide, Australia
| | - John R. Huguenard
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurology & Neurological Sciences, Stanford University; Stanford, CA, USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University; Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
9
|
Wang J, Meng N, Chen K, Huang X, Feng L, Yang C, Li Z, Sun X. The Relationship Between Depressive Symptoms and Functional Gastrointestinal Disorders (FGIDs): The Chain Mediating Effect of Sleep Disorders and Somatic Symptom. Depress Anxiety 2024; 2024:5586123. [PMID: 40226664 PMCID: PMC11918619 DOI: 10.1155/2024/5586123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/24/2024] [Accepted: 09/26/2024] [Indexed: 04/15/2025] Open
Abstract
Background: More than two-thirds of patients with functional gastrointestinal disorders (FGIDs) experience various degrees of mental health issues. Although studies indicate that FGIDs are related to depressive symptoms, sleep disorders, and somatic symptoms, the underlying mechanism between these variables remains unknown. Our objective was to establish a model that outlines the interactions between these psychological dimensions in FGIDs and, thus, provide valuable insights into how to enhance the well-being of affected individuals. Methods: This study used the convenient sampling method to enroll patients who visited the digestive internal medicine department. A total of 238 patients were investigated using the Rome IV criteria (irritable bowel syndrome used Rome Ⅲ criteria). A questionnaire including the Hospital Anxiety and Depressive Symptoms Scale, the Pittsburgh Sleep Quality Index, and the Patient Health Questionnaire-12 was used. The chain mediating roles of sleep disorders and somatic symptoms in the relationship between depressive symptoms and FGIDs were examined by the bootstrap method. Results: Correlation analysis revealed that depressive symptoms were positively related to sleep disorders, somatic symptoms, and FGIDs. Sleep disorders were positively related to somatic symptoms and FGIDs. Somatic symptoms were positively related to FGIDs. Chain mediating effect analysis showed that depressive symptoms can not only affect FGIDs but also through three indirect paths, as follows: the mediating role of sleep disorders and somatic symptoms, the chain mediating roles of sleep disorders and somatic symptoms, and the mediating effect size accounted for 7.2%, 7.7%, and 2.5% of the total effect, respectively. Conclusions: This study is conducive to understanding the internal mechanism underlying the relationship between depressive symptoms and FGIDs. It reminds us that when treating FGIDs patients, we should not only provide adequate psychological support to improve but also pay attention to improvements in their sleep quality and somatic symptoms.
Collapse
Affiliation(s)
- Jiana Wang
- School of Public Health, Health Science Center, Ningbo University, No. 818 Fenghua Road, Ningbo 315211, Zhejiang, China
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Nana Meng
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Kun Chen
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Xinyuan Huang
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Lin Feng
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Cong Yang
- Department of Social Medicine, School of Health Management, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang, Liaoning 110122, China
| | - Zhe Li
- Anesthesiology Department, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, No. 77 Puhe Road, Shenyang, North New Area, Shenyang 110122, Liaoning, China
| |
Collapse
|
10
|
Van Remoortel S, Hussein H, Boeckxstaens G. Mast cell modulation: A novel therapeutic strategy for abdominal pain in irritable bowel syndrome. Cell Rep Med 2024; 5:101780. [PMID: 39378882 PMCID: PMC11513802 DOI: 10.1016/j.xcrm.2024.101780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent gastrointestinal disorders characterized by recurrent abdominal pain and an altered defecation pattern. Chronic abdominal pain represents the hallmark IBS symptom and is reported to have the most bothersome impact on the patient's quality of life. Unfortunately, effective therapeutic strategies reducing abdominal pain are lacking, mainly attributed to a limited understanding of the contributing mechanisms. In the past few years, exciting new insights have pointed out that altered communication between gut immune cells and pain-sensing nerves acts as a hallmark driver of IBS-related abdominal pain. In this review, we aim to summarize our current knowledge on altered neuro-immune crosstalk as the main driver of altered pain signaling, with a specific focus on altered mast cell functioning herein, and highlight the relevance of targeting mast cell-mediated mechanisms as a novel therapeutic strategy for chronic abdominal pain in IBS patients.
Collapse
Affiliation(s)
- Samuel Van Remoortel
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Hind Hussein
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Centre for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium.
| |
Collapse
|
11
|
Fung C, Vanden Berghe P. Regenerating enteric neurites navigate the adult intestine using a glial positioning system? Neuron 2024; 112:2993-2995. [PMID: 39326387 DOI: 10.1016/j.neuron.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
While the enteric nervous system (ENS) is highly dynamic during development, the extent to which it is capable of repair remains unclear. In this issue of Neuron, Stavely et al.1 show that enteric neurons can reinnervate damaged regions to regain functionality using a glial positioning system (GPS) as their guide.
Collapse
Affiliation(s)
- Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Amedzrovi Agbesi RJ, El Merhie A, Spencer NJ, Hibberd T, Chevalier NR. Tetrodotoxin-resistant mechanosensitivity and L-type calcium channel-mediated spontaneous calcium activity in enteric neurons. Exp Physiol 2024; 109:1545-1556. [PMID: 38979869 PMCID: PMC11363105 DOI: 10.1113/ep091977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
Gut motility undergoes a switch from myogenic to neurogenic control in late embryonic development. Here, we report on the electrical events that underlie this transition in the enteric nervous system, using the GCaMP6f reporter in neural crest cell derivatives. We found that spontaneous calcium activity is tetrodotoxin (TTX) resistant at stage E11.5, but not at E18.5. Motility at E18.5 was characterized by periodic, alternating high- and low-frequency contractions of the circular smooth muscle; this frequency modulation was inhibited by TTX. Calcium imaging at the neurogenic-motility stages E18.5-P3 showed that CaV1.2-positive neurons exhibited spontaneous calcium activity, which was inhibited by nicardipine and 2-aminoethoxydiphenyl borate (2-APB). Our protocol locally prevented muscle tone relaxation, arguing for a direct effect of nicardipine on enteric neurons, rather than indirectly by its relaxing effect on muscle. We demonstrated that the ENS was mechanosensitive from early stages on (E14.5) and that this behaviour was TTX and 2-APB resistant. We extended our results on L-type channel-dependent spontaneous activity and TTX-resistant mechanosensitivity to the adult colon. Our results shed light on the critical transition from myogenic to neurogenic motility in the developing gut, as well as on the intriguing pathways mediating electro-mechanical sensitivity in the enteric nervous system. HIGHLIGHTS: What is the central question of this study? What are the first neural electric events underlying the transition from myogenic to neurogenic motility in the developing gut, what channels do they depend on, and does the enteric nervous system already exhibit mechanosensitivity? What is the main finding and its importance? ENS calcium activity is sensitive to tetrodotoxin at stage E18.5 but not E11.5. Spontaneous electric activity at fetal and adult stages is crucially dependent on L-type calcium channels and IP3R receptors, and the enteric nervous system exhibits a tetrodotoxin-resistant mechanosensitive response. Abstract figure legend Tetrodotoxin-resistant Ca2+ rise induced by mechanical stimulation in the E18.5 mouse duodenum.
Collapse
Affiliation(s)
| | - Amira El Merhie
- Laboratoire Matière et Systèmes Complexes UMR 7057Université Paris Cité/CNRSParisFrance
| | - Nick J. Spencer
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Tim Hibberd
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Nicolas R. Chevalier
- Laboratoire Matière et Systèmes Complexes UMR 7057Université Paris Cité/CNRSParisFrance
| |
Collapse
|
13
|
Gu W, Eke C, Gonzalez Santiago E, Olaloye O, Konnikova L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol 2024; 17:599-617. [PMID: 38555026 PMCID: PMC11384551 DOI: 10.1016/j.mucimm.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA; Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Voss U. Enteric neuroprotection-A matter of balancing redox potentials, limiting inflammation, and boosting resilience. Neurogastroenterol Motil 2024:e14871. [PMID: 39038122 DOI: 10.1111/nmo.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/15/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
The enteric nervous system (ENS) orchestrates intricate and autonomous functions throughout the gastrointestinal (GI) tract. Disruptions in ENS function are associated GI disorders. This mini review focuses on the past decade's research, utilizing rodent models, with an emphasis on protecting enteric neurons from loss. The review specifically looks at efforts to reduce oxidative stress, limit inflammation, and enhance neuronal resilience. Protective interventions including administration of antioxidants and compounds targeting cellular redox buffer systems, are evaluated for their effectiveness in preventing loss of enteric neurons in the ischemia-reperfusion model and streptozotocin-induced diabetes model. Interventions such as engrafting mesenchymal stem cells and targeting inflammatory signaling pathways in enteric neurons and glial cells are evaluated in inflammatory bowel disease models including the Winnie mouse, DSS-, and DNBS/TNBS-induced colitis models. The review also touches upon neuronal resilience, particularly in the context of Parkinson's disease models. Including estrogen's neuroprotective role, and the influence of metal ions on enteric neuronal protection. Understanding the dynamic interplay within the ENS and its role in disease pathogenesis holds promise for developing targeted therapies to effectively manage and treat various GI ailments.
Collapse
Affiliation(s)
- Ulrikke Voss
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Jamka JR, Gulbransen BD. Mechanisms of enteric neuropathy in diverse contexts of gastrointestinal dysfunction. Neurogastroenterol Motil 2024:e14870. [PMID: 39038157 DOI: 10.1111/nmo.14870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
The enteric nervous system (ENS) commands moment-to-moment gut functions through integrative neurocircuitry housed in the gut wall. The functional continuity of ENS networks is disrupted in enteric neuropathies and contributes to major disturbances in normal gut activities including abnormal gut motility, secretions, pain, immune dysregulation, and disrupted signaling along the gut-brain axis. The conditions under which enteric neuropathy occurs are diverse and the mechanistic underpinnings are incompletely understood. The purpose of this brief review is to summarize the current understanding of the cell types involved, the conditions in which neuropathy occurs, and the mechanisms implicated in enteric neuropathy such as oxidative stress, toll like receptor signaling, purines, and pre-programmed cell death.
Collapse
Affiliation(s)
- Julia R Jamka
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
16
|
Morys J, Małecki A, Nowacka-Chmielewska M. Stress and the gut-brain axis: an inflammatory perspective. Front Mol Neurosci 2024; 17:1415567. [PMID: 39092201 PMCID: PMC11292226 DOI: 10.3389/fnmol.2024.1415567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
The gut-brain axis (GBA) plays a dominant role in maintaining homeostasis as well as contributes to mental health maintenance. The pathways that underpin the axis expand from macroscopic interactions with the nervous system, to the molecular signals that include microbial metabolites, tight junction protein expression, or cytokines released during inflammation. The dysfunctional GBA has been repeatedly linked to the occurrence of anxiety- and depressive-like behaviors development. The importance of the inflammatory aspects of the altered GBA has recently been highlighted in the literature. Here we summarize current reports on GBA signaling which involves the immune response within the intestinal and blood-brain barrier (BBB). We also emphasize the effect of stress response on altering barriers' permeability, and the therapeutic potential of microbiota restoration by probiotic administration or microbiota transplantation, based on the latest animal studies. Most research performed on various stress models showed an association between anxiety- and depressive-like behaviors, dysbiosis of gut microbiota, and disruption of intestinal permeability with simultaneous changes in BBB integrity. It could be postulated that under stress conditions impaired communication across BBB may therefore represent a significant mechanism allowing the gut microbiota to affect brain functions.
Collapse
Affiliation(s)
| | | | - Marta Nowacka-Chmielewska
- Laboratory of Molecular Biology, Institute of Physiotherapy and Health Sciences, Academy of Physical Education, Katowice, Poland
| |
Collapse
|
17
|
McKay DM, Defaye M, Rajeev S, MacNaughton WK, Nasser Y, Sharkey KA. Neuroimmunophysiology of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2024; 326:G712-G725. [PMID: 38626403 PMCID: PMC11376980 DOI: 10.1152/ajpgi.00075.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/18/2024]
Abstract
Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.
Collapse
Affiliation(s)
- Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sruthi Rajeev
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
18
|
Suman S. Enteric Nervous System Alterations in Inflammatory Bowel Disease: Perspectives and Implications. GASTROINTESTINAL DISORDERS 2024; 6:368-379. [PMID: 38872954 PMCID: PMC11175598 DOI: 10.3390/gidisord6020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
The enteric nervous system (ENS), consisting of neurons and glial cells, is situated along the gastrointestinal (GI) tract's wall and plays a crucial role in coordinating digestive processes. Recent research suggests that the optimal functioning of the GI system relies on intricate connections between the ENS, the intestinal epithelium, the immune system, the intestinal microbiome, and the central nervous system (CNS). Inflammatory bowel disease (IBD) encompasses a group of chronic inflammatory disorders, such as Crohn's disease (CD) and ulcerative colitis (UC), characterized by recurring inflammation and damage to the GI tract. This review explores emerging research in the dynamic field of IBD and sheds light on the potential role of ENS alterations in both the etiology and management of IBD. Specifically, we delve into IBD-induced enteric glial cell (EGC) activation and its implications for persistent enteric gliosis, elucidating how this activation disrupts GI function through alterations in the gut-brain axis (GBA). Additionally, we examine IBD-associated ENS alterations, focusing on EGC senescence and the acquisition of the senescence-associated secretory phenotype (SASP). We highlight the pivotal role of these changes in persistent GI inflammation and the recurrence of IBD. Finally, we discuss potential therapeutic interventions involving senotherapeutic agents, providing insights into potential avenues for managing IBD by targeting ENS-related mechanisms. This approach might represent a potential alternative to managing IBD and advance treatment of this multifaceted disease.
Collapse
Affiliation(s)
- Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
19
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
20
|
Mukai H, Takanashi M, Ogawara KI, Maruyama M, Higaki K. Possible Regulation of P-Glycoprotein Function by Adrenergic Agonists II: Study with Isolated Rat Jejunal Sheets and Caco-2 Cell monolayers. J Pharm Sci 2024; 113:1209-1219. [PMID: 37984697 DOI: 10.1016/j.xphs.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
To clarify the regulation of drug absorption by the enteric nervous system, we investigated how adrenergic agonists (adrenaline (ADR), clonidine (CLO), dobutamine (DOB)) and dibutyryl cAMP (DBcAMP) affected P-glycoprotein (P-gp) function by utilizing isolated rat jejunal sheets and Caco-2 cell monolayers. ADR and CLO significantly decreased the secretory transport (Papptotal) of rhodamine-123 and tended to decrease the transport via P-gp (PappP-gp) and passive transport (Papppassive). In contrast, DBcAMP significantly increased and DOB tended to increase Papptotal and both tended to increase PappP-gpand Papppassive. Changes in P-gp expression on brush border membrane by adrenergic agonists and DBcAMP were significantly correlated with PappP-gp, while P-gp expression was not changed in whole cell homogenates, suggesting that the trafficking of P-gp would be responsible for its functional changes. Papppassive was inversely correlated with transmucosal or transepithelial electrical resistance, indicating that adrenergic agonists affected the paracellular permeability. Adrenergic agonists also changed cAMP levels, which were significantly correlated with PappP-gp. Furthermore, protein kinase A (PKA) or PKC inhibitor significantly decreased PappP-gp in Caco-2 cell monolayers, suggesting that they would partly contribute to the changes in P-gp activity. In conclusion, adrenergic agonists regulated P-gp function and paracellular permeability, which would be caused via adrenoceptor stimulation.
Collapse
Affiliation(s)
- Hironori Mukai
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Production Department, Odawara Central Factory, Nippon Shinyaku Co., Ltd., 676-1 Kuwahara, Odawara, Kanagawa 250-0861, Japan
| | - Masashi Takanashi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Central Hyogo Area, Hanshin Dispensing Pharmacy, I & H Co., Ltd., 1-18 Ohmasu-cho, Ashiya, Hyogo 659-0066, Japan
| | - Ken-Ichi Ogawara
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1, Motoyamakita, Higashinada-ku, Kobe, Hyogo 658-8558, Japan
| | - Masato Maruyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
21
|
Robertson K, Hahn O, Robinson BG, Faruk AT, Janakiraman M, Namkoong H, Kim K, Ye J, Bishop ES, Hall RA, Wyss-Coray T, Becker LS, Kaltschmidt JA. Gpr37 modulates the severity of inflammation-induced GI dysmotility by regulating enteric reactive gliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588619. [PMID: 38645163 PMCID: PMC11030428 DOI: 10.1101/2024.04.09.588619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The enteric nervous system (ENS) is contained within two layers of the gut wall and is made up of neurons, immune cells, and enteric glia cells (EGCs) that regulate gastrointestinal (GI) function. EGCs in both inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) change in response to inflammation, referred to as reactive gliosis. Whether EGCs restricted to a specific layer or region within the GI tract alone can influence intestinal immune response is unknown. Using bulk RNA-sequencing and in situ hybridization, we identify G-protein coupled receptor Gpr37 , as a gene expressed only in EGCs of the myenteric plexus, one of the two layers of the ENS. We show that Gpr37 contributes to key components of LPS-induced reactive gliosis including activation of NF-kB and IFN-y signaling and response genes, lymphocyte recruitment, and inflammation-induced GI dysmotility. Targeting Gpr37 in EGCs presents a potential avenue for modifying inflammatory processes in the ENS.
Collapse
|
22
|
Schneider S, Anderson JB, Bradley RP, Beigel K, Wright CM, Maguire BA, Yan G, Taylor DM, Harbour JW, Heuckeroth RO. BAP1 is required prenatally for differentiation and maintenance of postnatal murine enteric nervous system. J Clin Invest 2024; 134:e177771. [PMID: 38690732 PMCID: PMC11060734 DOI: 10.1172/jci177771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/05/2024] [Indexed: 05/03/2024] Open
Abstract
Epigenetic regulatory mechanisms are underappreciated, yet are critical for enteric nervous system (ENS) development and maintenance. We discovered that fetal loss of the epigenetic regulator Bap1 in the ENS lineage caused severe postnatal bowel dysfunction and early death in Tyrosinase-Cre Bap1fl/fl mice. Bap1-depleted ENS appeared normal in neonates; however, by P15, Bap1-deficient enteric neurons were largely absent from the small and large intestine of Tyrosinase-Cre Bap1fl/fl mice. Bowel motility became markedly abnormal with disproportionate loss of cholinergic neurons. Single-cell RNA sequencing at P5 showed that fetal Bap1 loss in Tyrosinase-Cre Bap1fl/fl mice markedly altered the composition and relative proportions of enteric neuron subtypes. In contrast, postnatal deletion of Bap1 did not cause enteric neuron loss or impaired bowel motility. These findings suggest that BAP1 is critical for postnatal enteric neuron differentiation and for early enteric neuron survival, a finding that may be relevant to the recently described human BAP1-associated neurodevelopmental disorder.
Collapse
Affiliation(s)
- Sabine Schneider
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica B. Anderson
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecca P. Bradley
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Katherine Beigel
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Christina M. Wright
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Beth A. Maguire
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Guang Yan
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Deanne M. Taylor
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - J. William Harbour
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert O. Heuckeroth
- Children’s Hospital of Philadelphia Research Institute, Abramson Research Center, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Delfino G, Briand JB, Oullier T, Nienkemper L, Greig J, Véziers J, Neunlist M, Derkinderen P, Paillusson S. Characterization of mitochondria-associated ER membranes in the enteric nervous system under physiological and pathological conditions. Am J Physiol Gastrointest Liver Physiol 2024; 326:G330-G343. [PMID: 38226933 PMCID: PMC11211041 DOI: 10.1152/ajpgi.00224.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/17/2024]
Abstract
Alterations in endoplasmic reticulum (ER)-mitochondria associations and in mitochondria-associated ER membrane (MAM) behavior have been reported in the brain in several neurodegenerative diseases. Despite the emerging role of the gut-brain axis in neurodegenerative disorders, the biology of MAM in the enteric nervous system (ENS) has not previously been studied. Therefore, we set out to characterize the MAM in the distal colon of wild-type C57BL/6J mice and senescence-accelerated mouse prone 8 (SAMP8), a mouse model of age-related neurodegeneration. We showed for the first time that MAMs are widely present in enteric neurons and that their association is altered in SAMP8 mice. We then examined the functions of MAMs in a primary culture model of enteric neurons and showed that calcium homeostasis was altered in SAMP8 mice when compared with control animals. These findings provide the first detailed characterization of MAMs in the ENS under physiological conditions and during age-associated neurodegeneration. Further investigation of MAM modifications in the ENS in disease may provide valuable information about the possible role of enteric MAMs in neurodegenerative diseases.NEW & NOTEWORTHY Our work shows for the first time the presence of contacts between endoplasmic reticulum and mitochondria in the enteric neurons and that the dynamic of these contacts is affected in these cells from an age-related neurodegeneration mouse model. It provides new insights into the potential role of enteric mitochondria-associated endoplasmic reticulum membrane in neurodegenerative disorders.
Collapse
Affiliation(s)
- Giada Delfino
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Jean Baptiste Briand
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Thibauld Oullier
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Léa Nienkemper
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Jenny Greig
- INSERM, Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie Néphrologie, Nantes, France
| | - Joëlle Véziers
- INSERM, Regenerative Medicine and Skeleton, Nantes Université, Oniris, Univ Angers, RMeS, Nantes, France
| | - Michel Neunlist
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Pascal Derkinderen
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| | - Sébastien Paillusson
- Nantes Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Nantes, The Enteric Nervous System in Gut and Brain Disorders, Nantes, France
| |
Collapse
|
24
|
Ortego-Isasa I, Ortega-Morán JF, Lozano H, Stieglitz T, Sánchez-Margallo FM, Usón-Gargallo J, Pagador JB, Ramos-Murguialday A. Colonic Electrical Stimulation for Chronic Constipation: A Perspective Review. Biomedicines 2024; 12:481. [PMID: 38540095 PMCID: PMC10967790 DOI: 10.3390/biomedicines12030481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 11/11/2024] Open
Abstract
Chronic constipation affects around 20% of the population and there is no efficient solution. This perspective review explores the potential of colonic electric stimulation (CES) using neural implants and methods of bioelectronic medicine as a therapeutic way to treat chronic constipation. The review covers the neurophysiology of colonic peristaltic function, the pathophysiology of chronic constipation, the technical aspects of CES, including stimulation parameters, electrode placement, and neuromodulation target selection, as well as a comprehensive analysis of various animal models highlighting their advantages and limitations in elucidating the mechanistic insights and translational relevance for CES. Finally, the main challenges and trends in CES are discussed.
Collapse
Affiliation(s)
- Iñaki Ortego-Isasa
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 San Sebastian, Spain; (H.L.); (A.R.-M.)
| | | | - Héctor Lozano
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 San Sebastian, Spain; (H.L.); (A.R.-M.)
| | - Thomas Stieglitz
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering–IMTEK and BrainLinks-BrainTools Center, University of Freiburg, 79110 Freiburg, Germany;
| | - Francisco M. Sánchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.F.O.-M.); (F.M.S.-M.); (J.U.-G.)
- TERAV/ISCIII, Red Española de Terapias Avanzadas, Instituto de Salud Carlos III (RICORS, RD21/0017/0029), 28029 Madrid, Spain
| | - Jesús Usón-Gargallo
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.F.O.-M.); (F.M.S.-M.); (J.U.-G.)
| | - J. Blas Pagador
- Jesús Usón Minimally Invasive Surgery Centre, 10071 Cáceres, Spain; (J.F.O.-M.); (F.M.S.-M.); (J.U.-G.)
- TERAV/ISCIII, Red Española de Terapias Avanzadas, Instituto de Salud Carlos III (RICORS, RD21/0017/0029), 28029 Madrid, Spain
| | - Ander Ramos-Murguialday
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 San Sebastian, Spain; (H.L.); (A.R.-M.)
- Department of Neurology and Stroke, University of Tubingen, 72076 Tubingen, Germany
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tubingen, 72076 Tubingen, Germany
- Athenea Neuroclinics, 20014 San Sebastian, Spain
| |
Collapse
|
25
|
Boesmans W. Fresh perspectives on how to build, maintain and repair the ENS. Nat Rev Gastroenterol Hepatol 2024; 21:82-83. [PMID: 37978241 DOI: 10.1038/s41575-023-00870-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Affiliation(s)
- Werend Boesmans
- Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium.
- Department of Pathology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
26
|
Idrizaj E, Nistri S, Nardini P, Baccari MC. Adiponectin affects ileal contractility of mouse preparations. Am J Physiol Gastrointest Liver Physiol 2024; 326:G187-G194. [PMID: 38111974 DOI: 10.1152/ajpgi.00203.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
Adiponectin (ADPN) has been reported to induce inhibitory effects on gastric motor activity, which, being a source of peripheral satiety signals, would contribute to the central anorexigenic effects of the hormone in rodents. However, peripheral satiety signals can also originate from the small intestine. Since there are no data on the effects of ADPN in this gut region, the present study aimed to investigate whether ADPN affects murine ileal contractility. Immunofluorescence experiments and Western blot were also performed to reveal the expression of ADPN receptors. Mechanical responses of ileal preparations were recorded in vitro via force-displacement transducers. Preparations showed a tetrodotoxin- and atropine-insensitive spontaneous contractile activity. Electrical field stimulation (EFS) induced tetrodotoxin- and atropine-sensitive contractile responses. ADPN induced a decay of the basal tension and decreased the amplitude of either the spontaneous contractility or the EFS-induced excitatory responses. All ADPN effects were abolished by the nitric oxide (NO) synthesis inhibitor NG-nitro l-arginine. The expression of the ADPN receptor, AdipoR1, but not AdipoR2, was also revealed in enteric glial cells. The present results offer the first evidence that ADPN acts on ileal preparations. The hormone exerts inhibitory effects, likely involving AdipoR1 on enteric glial cells and NO. From a physiological point of view, it could be hypothesized that the depressant action of ADPN on ileal contractility represents an additional peripheral satiety signal which, as also described for the ileal brake, could contribute to the central anorexigenic effects of the hormone.NEW & NOTEWORTHY This study provides the first evidence that adiponectin (ADPN) is able to act on ileal preparations. Functional results demonstrate that the hormone, other than causing a slight decay of the basal tension, depresses the amplitude of both spontaneous contractility and neurally induced excitatory responses of the mouse ileum through the involvement of nitric oxide. The expression of the ADPN receptor AdipoR1 and its localization on glial cells was revealed by Western blot and immunofluorescence analysis.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Firenze, Italy
| | - Silvia Nistri
- Department of Experimental and Clinical Medicine, Imaging Platform, University of Florence, Firenze, Italy
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, Imaging Platform, University of Florence, Firenze, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Firenze, Italy
| |
Collapse
|
27
|
Li J, Liu F, Mo K, Ni H, Yin Y. Effects of weaning on intestinal longitudinal muscle-myenteric plexus function in piglets. SCIENCE CHINA. LIFE SCIENCES 2024; 67:379-390. [PMID: 37824029 DOI: 10.1007/s11427-022-2391-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/18/2023] [Indexed: 10/13/2023]
Abstract
Weaning piglets usually suffer from severe diarrhea (commonly known as postweaning diarrhea, PWD) along with intestinal motility disorder. Intestinal peristalsis is mainly regulated by the longitudinal muscle-myenteric plexus (LM-MP). To understand the relationship between intestinal LM-MP function and the development of PWD, we compared the intestinal electrical activity, and the transcriptional profile of the LM-MP between 21-day-old piglets (just weaned, n=7) and 24-day-old piglets (suffered the most severe weaning stress, n=7). The results showed that 24-day-old piglets exhibited different degrees of diarrhea. A significant increase in the slow-wave frequency in the ileum and colon was observed in 24-day-old piglets, while c-kit expression in the intestinal LM-MPs was significantly decreased, indicating that PWD caused by elevated slow-wave frequency may be associated with loss of c-kit. The real-time quantitative PCR (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) showed that intestinal LM-MPs in 24-day-old piglets may undergo inflammation and oxidative stress. Significant increases in 8-hydroxy-2'-deoxyguanosine and decreases in thioredoxin suggest that weaning may lead to DNA damage in the LM-MP of 24-day-old piglets. In addition, activating transcription factor 3 was significantly upregulated, indicating nerve damage in the LM-MP of 24-day-old piglets. The transcriptomic results showed that most of the differentially expressed genes in the ileal LM-MP after weaning were downregulated and closely related to the cell cycle process. Subsequent RT-qPCR analysis showed that the relative expression of p21 was upregulated, while the expression of cyclin A2, cyclin B1, and proliferating cell nuclear antigen was downregulated in the ileal and colonic LM-MP of 24-day-old piglets, suggesting that weaning may inhibit cell proliferation and cause G1/S cell cycle arrest in ileal and colonic LM-MP. In conclusion, weaning may lead to cell cycle arrest by causing DNA damage in the LM-MP, impairing intestinal motility regulation, and ultimately leading to diarrhea in piglets.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Fenfen Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100008, China
| | - Kaibin Mo
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hengjia Ni
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 100008, China.
| |
Collapse
|
28
|
Zhao Y, Luo H, Ren X, Jia B, Li J, Wang L, Li J. The P2Y 1 receptor in the colonic myenteric plexus of rats and its correlation with opioid-induced constipation. BMC Gastroenterol 2024; 24:23. [PMID: 38191294 PMCID: PMC10773096 DOI: 10.1186/s12876-024-03119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/01/2024] [Indexed: 01/10/2024] Open
Abstract
This study was designed to explore the expression changes of P2Y1 receptors in the distal colonic myenteric layer of rats. An opioid induced constipation(OIC) rat model was generated by intraperitoneal (i.p) injection of loperamide. At 7 days post-treatment, the model rats were assessed by calculating the fecal water content and the gastrointestinal transit ratio. The immunofluorescence (IF)-based histochemical study was used to observe the distribution of P2Y1 receptors in the distal colonic myenteric plexus. Western blotting (WB) was performed to evaluate the expression changes of P2Y1 proteins in the myenteric layer, and the electrophysiological approaches were carried out to determine the regulatory roles of P2Y1 receptors on distal colonic motor function. IF showed that P2Y1 receptors are co-expressed MOR in the enteric nerve cells of the distal colonic myenteric plexus. Moreover, the WB revealed that the protein levels of P2Y1 were significantly decreased in the distal colonic myenteric layer of OIC rats. In vitro tension experiments exhibited that the P2Y1 receptor antagonist MRS2500 enhanced the spontaneous contraction amplitude, adding EM2 and β-FNA did not have any effect on MRS2500. Therefore, P2Y1 receptor expression could be associated with the occurrence of OIC in this rat model and the regulation of colonic motility by MOR may be related to the release of purine neurotransmitters such as ATP in the colonic nervous system.
Collapse
Affiliation(s)
- Yuqiong Zhao
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Huijuan Luo
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Xiaojie Ren
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Binghan Jia
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Jinzhao Li
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China
| | - Lixin Wang
- The Medical Laboratory Center of General Hospital of Ningxia Medical University, 804 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China.
| | - Junping Li
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, 750001, Yinchuan, Ningxia Hui Autonomous Region, P.R. China.
| |
Collapse
|
29
|
Kardan R, Hemmati J, Nazari M, Ahmadi A, Asghari B, Azizi M, Khaledi M, Arabestani MR. Novel therapeutic strategy for obesity through the gut microbiota-brain axis: A review article. CASPIAN JOURNAL OF INTERNAL MEDICINE 2024; 15:215-227. [PMID: 38807723 PMCID: PMC11129059 DOI: 10.22088/cjim.15.2.215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/27/2023] [Accepted: 05/27/2023] [Indexed: 05/30/2024]
Abstract
Background: The interaction between commensal bacteria and the host is essential for health and the gut microbiota-brain axis plays a vital role in this regard. Obesity as a medical problem not only affect the health of the individuals, but also the economic and social aspects of communities. The presence of any dysbiosis in the composition of the gut microbiota disrupts in the gut microbiota-brain axis, which in turn leads to an increase in appetite and then obesity. Because common treatments for obesity have several drawbacks, the use of microbiota-based therapy in addition to treatment and prevention of obesity can have other numerous benefits for the individual. In this review, we intend to investigate the relationship between obesity and the gut microbiota-brain axis as well as novel treatment strategies based on this axis with an emphasis on gut microbiota.
Collapse
Affiliation(s)
- Romina Kardan
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- These authors contributed equally in this article
| | - Jaber Hemmati
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- These authors contributed equally in this article
| | - Mohsen Nazari
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amjad Ahmadi
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Babak Asghari
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mansoor Khaledi
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Reza Arabestani
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
30
|
Gomez-Frittelli J, Hamnett R, Kaltschmidt JA. Comparison of wholemount dissection methods for neuronal subtype marker expression in the mouse myenteric plexus. Neurogastroenterol Motil 2024; 36:e14693. [PMID: 37882149 PMCID: PMC10842488 DOI: 10.1111/nmo.14693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 09/05/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Accurately reporting the identity and representation of enteric nervous system (ENS) neuronal subtypes along the length of the gastrointestinal (GI) tract is critical to advancing our understanding of ENS control of GI function. Reports of varying proportions of subtype marker expression have employed different dissection techniques to achieve wholemount muscularis preparations of myenteric plexus. In this study, we asked whether differences in GI dissection methods could introduce variability into the quantification of marker expression. METHODS We compared three commonly used methods of ENS wholemount dissection: two flat-sheet preparations that differed in the order of microdissection and fixation and a third rod-mounted peeling technique. We also tested a reversed orientation variation of flat-sheet peeling, two step-by-step variations of the rod peeling technique, and whole-gut fixation as a tube. We assessed marker expression using immunohistochemistry, genetic reporter lines, confocal microscopy, and automated image analysis. KEY RESULTS AND CONCLUSIONS We found no significant differences between the two flat-sheet preparation methods in the expression of calretinin or neuronal nitric oxide synthase (nNOS) as a proportion of total neurons in ileum myenteric plexus. However, the rod-mounted peeling method resulted in decreased proportion of neurons labeled for both calretinin and nNOS. This method also resulted in decreased transgenic reporter fluorescent protein (tdTomato) for substance P in distal colon and choline acetyltransferase (ChAT) in both ileum and distal colon. These results suggest that labeling among some markers, both native protein and transgenic fluorescent reporters, is decreased by the rod-mounted mechanical method of peeling. The step-by-step variations of this method point to mechanical manipulation of the tissue as the likely cause of decreased labeling. Our study thereby demonstrates a critical variability in wholemount muscularis dissection methods.
Collapse
Affiliation(s)
- Julieta Gomez-Frittelli
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305 USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305 USA
| | - Ryan Hamnett
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305 USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305 USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305 USA
| |
Collapse
|
31
|
Liang F, Liu S, Zhang H, Xiang R, Xie M, He X, Wang S, Wu S, Li J. Effects of chronic unpredictable mild stress on gut sensation and function in male mice. Stress 2024; 27:2374768. [PMID: 38975691 DOI: 10.1080/10253890.2024.2374768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Stress has been linked to the development of irritable bowel syndrome (IBS), and various methods have been explored to model IBS in combination with other stimuli. However, it remains unclear whether stress alone can induce IBS in animals. This study aimed to investigate the impact of chronic unpredictable mild stress (CUMS) on gastrointestinal sensation and function in mice and assess the potential of CUMS as a modeling approach for IBS. To evaluate the mice's behavior, we conducted open field test, sucrose preference test and weighed the mice, revealing that CUMS indeed induced anxiety and depression in the mice and caused weight loss. Further analyses, including fecal analysis, a total gastrointestinal transport test, and a colon propulsion test, demonstrated that CUMS led to abnormal defecation and disruptions in gastrointestinal motility in the mice. Additionally, the abdominal withdrawal reflex test indicated an increase in visceral sensitivity in CUMS-exposed mice. Histological examination using hematoxylin and eosin staining revealed no significant histological alterations in the colons of CUMS-exposed mice, but it did show a minor degree of inflammatory cell infiltration. In summary, the findings suggest that CUMS can replicate IBS-like symptoms in mice, offering a novel top-down approach to modeling IBS.
Collapse
Affiliation(s)
- Fangyuan Liang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Suzhen Liu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Heng Zhang
- Department of Traditional Chinese Medicine, Henan Workers' Hospital, Zhengzhou, China
| | - Ronglan Xiang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Mengting Xie
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaoru He
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Sunyi Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Song Wu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Acupuncture and Moxibustion Department, Affiliated Hospital of Hubei University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Jia Li
- Acupuncture and Moxibustion, Xianning Hospital of Traditional Chinese Medicine Department, Xianning, China
| |
Collapse
|
32
|
Sasidharan A, Peethambar BA, Kumar KS, Kumar AV, Hiregange A, Fawkes N, Collins JF, Grosche A, Vidyasagar S. Advancing peristalsis deciphering in mouse small intestine by multi-parameter tracking. Commun Biol 2023; 6:1237. [PMID: 38062160 PMCID: PMC10703907 DOI: 10.1038/s42003-023-05631-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Assessing gastrointestinal motility lacks simultaneous evaluation of intraluminal pressure (ILP), circular muscle (CM) and longitudinal muscle (LM) contraction, and lumen emptying. In this study, a sophisticated machine was developed that synchronized real-time recordings to quantify the intricate interplay between CM and LM contractions, and their timings for volume changes using high-resolution cameras with machine learning capability, the ILP using pressure transducers and droplet discharge (DD) using droplet counters. Results revealed four distinct phases, BPhase, NPhase, DPhase, and APhase, distinguished by pressure wave amplitudes. Fluid filling impacted LM strength and contraction frequency initially, followed by CM contraction affecting ILP, volume, and the extent of anterograde, retrograde, and segmental contractions during these phases that result in short or long duration DD. This comprehensive analysis sheds light on peristalsis mechanisms, understand their sequence and how one parameter influenced the other, offering insights for managing peristalsis by regulating smooth muscle contractions.
Collapse
Affiliation(s)
- Anusree Sasidharan
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | | | | - Ashok V Kumar
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, USA
| | | | | | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - Astrid Grosche
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
33
|
Zhao C, Zhou X, Shi X. The influence of Nav1.9 channels on intestinal hyperpathia and dysmotility. Channels (Austin) 2023; 17:2212350. [PMID: 37186898 DOI: 10.1080/19336950.2023.2212350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
The Nav1.9 channel is a voltage-gated sodium channel. It plays a vital role in the generation of pain and the formation of neuronal hyperexcitability after inflammation. It is highly expressed in small diameter neurons of dorsal root ganglions and Dogiel II neurons in enteric nervous system. The small diameter neurons in dorsal root ganglions are the primary sensory neurons of pain conduction. Nav1.9 channels also participate in regulating intestinal motility. Functional enhancements of Nav1.9 channels to a certain extent lead to hyperexcitability of small diameter dorsal root ganglion neurons. The hyperexcitability of the neurons can cause visceral hyperalgesia. Intestinofugal afferent neurons and intrinsic primary afferent neurons in enteric nervous system belong to Dogiel type II neurons. Their excitability can also be regulated by Nav1.9 channels. The hyperexcitability of intestinofugal afferent neurons abnormally activate entero-enteric inhibitory reflexes. The hyperexcitability of intrinsic primary afferent neurons disturb peristaltic waves by abnormally activating peristaltic reflexes. This review discusses the role of Nav1.9 channels in intestinal hyperpathia and dysmotility.
Collapse
Affiliation(s)
- Chenyu Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoliu Shi
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Liu Z, Chen X, Chen P, Wang L. Transcranial Direct Current Stimulation Attenuates the Chronic Pain of Osteoarthritis in Rats via Reducing NMDAR2B Expressions in the Spinal Cord. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2023; 23:498-505. [PMID: 38037367 PMCID: PMC10696371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Accepted: 10/29/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVES Osteoarthritis (OA) has been the common cause to lead to chronic pain. Transcranial direct current stimulation (tDCS) is effective in the treatment of chronic pain, but its analgesic mechanism is still unclear. This study observed the analgesic effects of tDCS in rats to explore the top-down analgesic modulation mechanism of tDCS. METHODS Monosodium iodoacetate (MIA) was used to establish OA chronic pain model. After 21 days, the rats received tDCS for 14 consecutive days (20 min/day). We assessed the pain-related behaviors of rats at different time points. Western blot and Immunohistochemistry were performed to observe the expression level of NMDAR2B in the spinal cord after tDCS treatment. RESULTS After MIA injection, rats developed apparent mechanical hyperalgesia and thermal hyperalgesia. However, the pain-related behaviors of rats were significantly improved after tDCS treatment. In addition, the expression of NMDAR2B and the proportion of positive stained cells of NMDAR2B were reversed by tDCS treatment. CONCLUSIONS The results demonstrated that tDCS can attenuate OA-induced chronic pain in rats via reducing NMDAR2B expressions in the spinal cord. We believe that this may be the result of tDCS participating in the top-down modulation of pain pathway in the endogenous analgesic system.
Collapse
Affiliation(s)
- Zhihua Liu
- Department of Rehabilitation Medicine, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong Province, China
| | - Xia Chen
- Department of Pulmonary and Critial Care Medicine (PCCM), Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong Province, China
| | - Peng Chen
- Department of Rehabilitation Medicine, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong Province, China
| | - Lili Wang
- Department of Pulmonary and Critial Care Medicine (PCCM), Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong Province, China
| |
Collapse
|
35
|
Zhao M, Wang P, Sun X, Yang D, Zhang S, Meng X, Zhang M, Gao X. Detrimental Impacts of Pharmaceutical Excipient PEG400 on Gut Microbiota and Metabolome in Healthy Mice. Molecules 2023; 28:7562. [PMID: 38005284 PMCID: PMC10673170 DOI: 10.3390/molecules28227562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Polyethylene glycol 400 (PEG400) is a widely used pharmaceutical excipient in the field of medicine. It not only enhances the dispersion stability of the main drug but also facilitates the absorption of multiple drugs. Our previous study found that the long-term application of PEG400 as an adjuvant in traditional Chinese medicine preparations resulted in wasting and weight loss in animals, which aroused our concern. In this study, 16S rRNA high-throughput sequencing technology was used to analyze the diversity of gut microbiota, and LC-MS/MS Q-Exactive Orbtriap metabolomics technology was used to analyze the effect of PEG400 on the metabolome of healthy mice, combined with intestinal pathological analysis, aiming to investigate the effects of PEG400 on healthy mice. These results showed that PEG400 significantly altered the structure of gut microbiota, reduced the richness and diversity of intestinal flora, greatly increased the abundance of Akkermansia muciniphila (A. muciniphila), increased the proportion of Bacteroidetes to Firmicutes, and reduced the abundance of many beneficial bacteria. Moreover, PEG400 changed the characteristics of fecal metabolome in mice and induced disorders in lipid and energy metabolism, thus leading to diarrhea, weight loss, and intestinal inflammation in mice. Collectively, these findings provide new evidence for the potential effect of PEG400 ingestion on a healthy host.
Collapse
Affiliation(s)
- Mei Zhao
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China;
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| | - Pengjiao Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| | - Xiaodong Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| | - Dan Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| | - Shuo Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
- Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Xiaoxia Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
- School of Medicine and Health Management, Guizhou Medical University, Guiyang 550025, China
| | - Min Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| | - Xiuli Gao
- School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China;
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (P.W.); (X.S.); (D.Y.); (S.Z.); (X.M.)
- Microbiology and Biochemical Pharmaceutical Engineering Research Center, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
36
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-related molecule Cntnap2 affects colonic motility in mice. Front Neurosci 2023; 17:1287057. [PMID: 38027494 PMCID: PMC10665486 DOI: 10.3389/fnins.2023.1287057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in vivo and ex vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV, United States
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
37
|
Cao Y, Chen H, Yang J. Neuroanatomy of lymphoid organs: Lessons learned from whole-tissue imaging studies. Eur J Immunol 2023; 53:e2250136. [PMID: 37377338 DOI: 10.1002/eji.202250136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Decades of extensive research have documented the presence of neural innervations of sensory, sympathetic, or parasympathetic origin in primary and secondary lymphoid organs. Such neural inputs can release neurotransmitters and neuropeptides to directly modulate the functions of various immune cells, which represents one of the essential aspects of the body's neuroimmune network. Notably, recent studies empowered by state-of-the-art imaging techniques have comprehensively assessed neural distribution patterns in BM, thymus, spleen, and LNs of rodents and humans, helping clarify several controversies lingering in the field. In addition, it has become evident that neural innervations in lymphoid organs are not static but undergo alterations in pathophysiological contexts. This review aims to update the current information on the neuroanatomy of lymphoid organs obtained through whole-tissue 3D imaging and genetic approaches, focusing on anatomical features that may designate the functional modulation of immune responses. Moreover, we discuss several critical questions that call for future research, which will advance our in-depth understanding of the importance and complexity of neural control of lymphoid organs.
Collapse
Affiliation(s)
- Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Hongjie Chen
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jing Yang
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, China
| |
Collapse
|
38
|
Yadav SK, Ito K, Dhib-Jalbut S. Interaction of the Gut Microbiome and Immunity in Multiple Sclerosis: Impact of Diet and Immune Therapy. Int J Mol Sci 2023; 24:14756. [PMID: 37834203 PMCID: PMC10572709 DOI: 10.3390/ijms241914756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The bidirectional communication between the gut and central nervous system (CNS) through microbiota is known as the microbiota-gut-brain axis. The brain, through the enteric neural innervation and the vagus nerve, influences the gut physiological activities (motility, mucin, and peptide secretion), as well as the development of the mucosal immune system. Conversely, the gut can influence the CNS via intestinal microbiota, its metabolites, and gut-homing immune cells. Growing evidence suggests that gut immunity is critically involved in gut-brain communication during health and diseases, including multiple sclerosis (MS). The gut microbiota can influence the development and function of gut immunity, and conversely, the innate and adaptive mucosal immunity can influence microbiota composition. Gut and systemic immunity, along with gut microbiota, are perturbed in MS. Diet and disease-modifying therapies (DMTs) can affect the composition of the gut microbial community, leading to changes in gut and peripheral immunity, which ultimately affects MS. A high-fat diet is highly associated with gut dysbiosis-mediated inflammation and intestinal permeability, while a high-fiber diet/short-chain fatty acids (SCFAs) can promote the development of Foxp3 Tregs and improvement in intestinal barrier function, which subsequently suppress CNS autoimmunity in the animal model of MS (experimental autoimmune encephalomyelitis or EAE). This review will address the role of gut immunity and its modulation by diet and DMTs via gut microbiota during MS pathophysiology.
Collapse
Affiliation(s)
- Sudhir Kumar Yadav
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (K.I.)
| | - Kouichi Ito
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (K.I.)
| | - Suhayl Dhib-Jalbut
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA; (S.K.Y.); (K.I.)
- Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| |
Collapse
|
39
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-Related Molecule Cntnap2 Affects Colonic Motility in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537221. [PMID: 37131706 PMCID: PMC10153124 DOI: 10.1101/2023.04.17.537221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in-vivo and ex-vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV 89557, USA
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
40
|
Mahapatra A, Dhakal A, Noguchi A, Vadlamani P, Hundley HA. ADAR-mediated regulation of PQM-1 expression in neurons impacts gene expression throughout C. elegans and regulates survival from hypoxia. PLoS Biol 2023; 21:e3002150. [PMID: 37747897 PMCID: PMC10553819 DOI: 10.1371/journal.pbio.3002150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/05/2023] [Accepted: 08/23/2023] [Indexed: 09/27/2023] Open
Abstract
The ability to alter gene expression programs in response to changes in environmental conditions is central to the ability of an organism to thrive. For most organisms, the nervous system serves as the master regulator in communicating information about the animal's surroundings to other tissues. The information relay centers on signaling pathways that cue transcription factors in a given cell type to execute a specific gene expression program, but also provide a means to signal between tissues. The transcription factor PQM-1 is an important mediator of the insulin signaling pathway contributing to longevity and the stress response as well as impacting survival from hypoxia. Herein, we reveal a novel mechanism for regulating PQM-1 expression specifically in neural cells of larval animals. Our studies reveal that the RNA-binding protein (RBP), ADR-1, binds to pqm-1 mRNA in neural cells. This binding is regulated by the presence of a second RBP, ADR-2, which when absent leads to reduced expression of both pqm-1 and downstream PQM-1 activated genes. Interestingly, we find that neural pqm-1 expression is sufficient to impact gene expression throughout the animal and affect survival from hypoxia, phenotypes that we also observe in adr mutant animals. Together, these studies reveal an important posttranscriptional gene regulatory mechanism in Caenorhabditis elegans that allows the nervous system to sense and respond to environmental conditions to promote organismal survival from hypoxia.
Collapse
Affiliation(s)
- Ananya Mahapatra
- Genome, Cell and Developmental Biology Graduate Program, Indiana University, Bloomington, Indiana, United States of America
| | - Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine–Bloomington, Bloomington, Indiana, United States of America
| | - Aika Noguchi
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| | - Pranathi Vadlamani
- Medical Sciences Program, Indiana University School of Medicine–Bloomington, Bloomington, Indiana, United States of America
| | - Heather A. Hundley
- Department of Biology, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
41
|
Shaffer C, Barrett LF, Quigley KS. Signal processing in the vagus nerve: Hypotheses based on new genetic and anatomical evidence. Biol Psychol 2023; 182:108626. [PMID: 37419401 PMCID: PMC10563766 DOI: 10.1016/j.biopsycho.2023.108626] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Each organism must regulate its internal state in a metabolically efficient way as it interacts in space and time with an ever-changing and only partly predictable world. Success in this endeavor is largely determined by the ongoing communication between brain and body, and the vagus nerve is a crucial structure in that dialogue. In this review, we introduce the novel hypothesis that the afferent vagus nerve is engaged in signal processing rather than just signal relay. New genetic and structural evidence of vagal afferent fiber anatomy motivates two hypotheses: (1) that sensory signals informing on the physiological state of the body compute both spatial and temporal viscerosensory features as they ascend the vagus nerve, following patterns found in other sensory architectures, such as the visual and olfactory systems; and (2) that ascending and descending signals modulate one another, calling into question the strict segregation of sensory and motor signals, respectively. Finally, we discuss several implications of our two hypotheses for understanding the role of viscerosensory signal processing in predictive energy regulation (i.e., allostasis) as well as the role of metabolic signals in memory and in disorders of prediction (e.g., mood disorders).
Collapse
Affiliation(s)
- Clare Shaffer
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA.
| | - Lisa Feldman Barrett
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Karen S Quigley
- Department of Psychology, College of Science, Northeastern University, Boston, MA, USA.
| |
Collapse
|
42
|
Lee CYQ, Balasuriya GK, Herath M, Franks AE, Hill-Yardin EL. Impaired cecal motility and secretion alongside expansion of gut-associated lymphoid tissue in the Nlgn3 R451C mouse model of autism. Sci Rep 2023; 13:12687. [PMID: 37542090 PMCID: PMC10403596 DOI: 10.1038/s41598-023-39555-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/27/2023] [Indexed: 08/06/2023] Open
Abstract
Individuals with Autism Spectrum Disorder (ASD; autism) commonly present with gastrointestinal (GI) illness in addition to core diagnostic behavioural traits. The appendix, or cecum in mice, is important for GI homeostasis via its function as a key site for fermentation and a microbial reservoir. Even so, the role of the appendix and cecum in autism-associated GI symptoms remains uninvestigated. Here, we studied mice with an autism-associated missense mutation in the post-synaptic protein neuroligin-3 (Nlgn3R451C), which impacts brain and enteric neuronal activity. We assessed for changes in cecal motility using a tri-cannulation video-imaging approach in ex vivo preparations from wild-type and Nlgn3R451C mice. We investigated cecal permeability and neurally-evoked secretion in wild-type and Nlgn3R451C tissues using an Ussing chamber set-up. The number of cecal patches in fresh tissue samples were assessed and key immune populations including gut macrophages and dendritic cells were visualised using immunofluorescence. Nlgn3R451C mice displayed accelerated cecal motor complexes and reduced cecal weight in comparison to wildtype littermates. Nlgn3R451C mice also demonstrated reduced neurally-evoked cecal secretion in response to the nicotinic acetylcholine receptor agonist 1,1-dimethyl-4-phenylpiperazinium (DMPP), but permeability was unchanged. We observed an increase in the number of cecal patches in Nlgn3R451C mice, however the cellular morphologies of key immune populations studied were not significantly altered. We show that the R451C nervous system mutation leads to cecal dysmotility, impaired secretion, and neuro-immune alterations. Together, these results suggest that the R451C mutation disrupts the gut-brain axis with GI dysfunction in autism.
Collapse
Affiliation(s)
- Chalystha Yie Qin Lee
- School of Health and Biomedical Sciences, RMIT University, 223, Bundoora West Campus, 225-245 Clements Drive, Bundoora, VIC, 3083, Australia
| | | | - Madushani Herath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX, USA
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Ashley E Franks
- School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, 223, Bundoora West Campus, 225-245 Clements Drive, Bundoora, VIC, 3083, Australia.
- Department of Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
43
|
Wolniczak E, Meyer F, Albrecht A. [The abdominal brain: neuroanatomic perspectives for the abdominal surgeon]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2023; 61:1037-1045. [PMID: 37142237 DOI: 10.1055/a-2013-7633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The "abdominal brain" does not only consist of a separate enteric nervous system but also of bidirectional connections to the autonomous nerve system with parasympathicus und sympathicus as well as brain and spinal cord. Novel studies have shown that these connections can quickly transfer information on the ingested nutrients to the brain to conduct the feeling of hunger and more complex behaviour, such as "reward-related learning". However, even emotional experience, in particular, stress, has a strong impact onto the gastrointestinal system. The immune system, motility and barrier function of the gastrointestinal tract are modulated by the intestinal microbiota. Local bacteria may directly influence neuronal communication by released metabolic products and neuropeptides as well as may control inflammatory factors. Intensive research over the last 10 years was able to provide evidence that intestinal microbiota may affect emotional and cognitive aspects of our behaviour and, thus, it might be in the focus of numerous neuropsychiatric diseases, such as depressions and anxiety disorders.The presented review is to provide a short summary of the I): anatomic basics of the so-called gut-brain axis and II): modi of the bidirectional regulation. Through indirect connections to the limbic system, gut-brain axis can substantially influence stress and anxiety but also the pain processing. In addition, the role of microbiota is outlined and future paths are shown, e.g., how the (microbiota-)gut-brain axis may alter emotional experience, pain processing and intestinal function. Such associations are relevant for further development of visceral medicine, and, thus, also for the abdominal surgeon to derive future treatment concepts with interdisciplinary orientation.
Collapse
Affiliation(s)
- Erik Wolniczak
- Institut für Anatomie, Otto-von-Guericke-Universität zu Magdeburg, Magdeburg, Deutschland
| | - Frank Meyer
- Klinik für Allgemein-, Viszeral-, Gefäß- und Transplantationschirurgie, Universitätsklinikum Magdeburg A.ö.R., Magdeburg, Deutschland
| | - Anne Albrecht
- Institut für Anatomie, Otto-von-Guericke-Universität zu Magdeburg, Magdeburg, Deutschland
- Center for Behavioral Brain Science (CBBS), Magdeburg, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
44
|
Herath M, Cho E, Marklund U, Franks AE, Bornstein JC, Hill-Yardin EL. Quantitative Spatial Analysis of Neuroligin-3 mRNA Expression in the Enteric Nervous System Reveals a Potential Role in Neuronal-Glial Synapses and Reduced Expression in Nlgn3R451C Mice. Biomolecules 2023; 13:1063. [PMID: 37509099 PMCID: PMC10377306 DOI: 10.3390/biom13071063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Mutations in the Neuroligin-3 (Nlgn3) gene are implicated in autism spectrum disorder (ASD) and gastrointestinal (GI) dysfunction, but cellular Nlgn3 expression in the enteric nervous system remains to be characterised. We combined RNAScope in situ hybridization and immunofluorescence to measure Nlgn3 mRNA expression in cholinergic and VIP-expressing submucosal neurons, nitrergic and calretinin-containing myenteric neurons and glial cells in both WT and Nlgn3R451C mutant mice. We measured Nlgn3 mRNA neuronal and glial expression via quantitative three-dimensional image analysis. To validate dual RNAScope/immunofluorescence data, we interrogated available single-cell RNA sequencing (scRNASeq) data to assess for Nlgn3, Nlgn1, Nlgn2 and their binding partners, Nrxn1-3, MGDA1 and MGDA2, in enteric neural subsets. Most submucosal and myenteric neurons expressed Nlgn3 mRNA. In contrast to other Nlgns and binding partners, Nlgn3 was strongly expressed in enteric glia, suggesting a role for neuroligin-3 in mediating enteric neuron-glia interactions. The autism-associated R451C mutation reduces Nlgn3 mRNA expression in cholinergic but not in VIPergic submucosal neurons. In the myenteric plexus, Nlgn3 mRNA levels are reduced in calretinin, nNOS-labelled neurons and S100 β -labelled glia. We provide a comprehensive cellular profile for neuroligin-3 expression in ileal neuronal subpopulations of mice expressing the R451C autism-associated mutation in Nlgn3, which may contribute to the understanding of the pathophysiology of GI dysfunction in ASD.
Collapse
Affiliation(s)
- Madushani Herath
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ulrika Marklund
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Ashley E Franks
- Department of Microbiology, Anatomy Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia
| | - Joel C Bornstein
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC 3083, Australia
| |
Collapse
|
45
|
Majd A, Richter MN, Samuel RM, Cesiulis A, Ghazizadeh Z, Wang J, Fattahi F. Combined GWAS and single cell transcriptomics uncover the underlying genes and cell types in disorders of gut-brain interaction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.02.23290906. [PMID: 37333423 PMCID: PMC10275016 DOI: 10.1101/2023.06.02.23290906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Disorders of gut-brain interaction (DGBIs), formerly known as functional gastrointestinal disorders, are extremely common and historically difficult to manage. This is largely because their cellular and molecular mechanisms have remained poorly understood and understudied. One approach to unravel the molecular underpinnings of complex disorders such as DGBIs is performing genome wide association studies (GWASs). However, due to the heterogenous and non-specific nature of GI symptoms, it has been difficult to accurately classify cases and controls. Thus, to perform reliable studies, we need to access large patient populations which has been difficult to date. Here, we leveraged the UK Biobank (UKBB) database, containing genetic and medical record data of over half a million individuals, to perform GWAS for five DGBI categories: functional chest pain, functional diarrhea, functional dyspepsia, functional dysphagia, and functional fecal incontinence. By applying strict inclusion and exclusion criteria, we resolved patient populations and identified genes significantly associated with each condition. Leveraging multiple human single-cell RNA-sequencing datasets, we found that the disease associated genes were highly expressed in enteric neurons, which innervate and control GI functions. Further expression and association testing-based analyses revealed specific enteric neuron subtypes consistently linked with each DGBI. Furthermore, protein-protein interaction analysis of each of the disease associated genes revealed protein networks specific to each DGBI, including hedgehog signaling for functional chest pain and neuronal function and neurotransmission for functional diarrhea and functional dyspepsia. Finally, through retrospective medical record analysis we found that drugs that inhibit these networks are associated with an increased disease risk, including serine/threonine kinase 32B drugs for functional chest pain, solute carrier organic anion transporter family member 4C1, mitogen-activated protein kinase 6, and dual serine/threonine and tyrosine protein kinase drugs for functional dyspepsia, and serotonin transporter drugs for functional diarrhea. This study presents a robust strategy for uncovering the tissues, cell types, and genes involved in DGBIs, presenting novel predictions of the mechanisms underlying these historically intractable and poorly understood diseases.
Collapse
Affiliation(s)
- Alireza Majd
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Mikayla N Richter
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Ryan M Samuel
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Andrius Cesiulis
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Zaniar Ghazizadeh
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Jeffrey Wang
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California, USA
- Program in Craniofacial Biology, University of California, San Francisco, California, USA
| |
Collapse
|
46
|
Wang L, Yuan PQ, Taché Y. Vasculature in the mouse colon and spatial relationships with the enteric nervous system, glia, and immune cells. Front Neuroanat 2023; 17:1130169. [PMID: 37332321 PMCID: PMC10272736 DOI: 10.3389/fnana.2023.1130169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/15/2023] [Indexed: 06/20/2023] Open
Abstract
The distribution, morphology, and innervation of vasculature in different mouse colonic segments and layers, as well as spatial relationships of the vasculature with the enteric plexuses, glia, and macrophages are far from being complete. The vessels in the adult mouse colon were stained by the cardiovascular perfusion of wheat germ agglutinin (WGA)-Alexa Fluor 448 and by CD31 immunoreactivity. Nerve fibers, enteric glia, and macrophages were immunostained in the WGA-perfused colon. The blood vessels entered from the mesentery to the submucosa and branched into the capillary networks in the mucosa and muscularis externa. The capillary net formed anastomosed rings at the orifices of mucosa crypts, and the capillary rings surrounded the crypts individually in the proximal colon and more than two crypts in the distal colon. Microvessels in the muscularis externa with myenteric plexus were less dense than in the mucosa and formed loops. In the circular smooth muscle layer, microvessels were distributed in the proximal, but not the distal colon. Capillaries did not enter the enteric ganglia. There were no significant differences in microvascular volume per tissue volume between the proximal and distal colon either in the mucosa or muscularis externa containing the myenteric plexus. PGP9.5-, tyrosine hydroxylase-, and calcitonin gene-related peptide (CGRP)-immunoreactive nerve fibers were distributed along the vessels in the submucosa. In the mucosa, PGP9.5-, CGRP-, and vasoactive intestinal peptide (VIP)-immunoreactive nerves terminated close to the capillary rings, while cells and processes labeled by S100B and glial fibrillary acidic protein were distributed mainly in the lamina propria and lower portion of the mucosa. Dense Iba1 immunoreactive macrophages were closely adjacent to the mucosal capillary rings. There were a few macrophages, but no glia in apposition to microvessels in the submucosa and muscularis externa. In conclusion, in the mouse colon, (1) the differences in vasculature between the proximal and distal colon were associated with the morphology, but not the microvascular amount per tissue volume in the mucosa and muscle layers; (2) the colonic mucosa contained significantly more microvessels than the muscularis externa; and (3) there were more CGRP and VIP nerve fibers found close to microvessels in the mucosa and submucosa than in the muscle layers.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Pu-Qing Yuan
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Yvette Taché
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
47
|
Viola MF, Chavero-Pieres M, Modave E, Delfini M, Stakenborg N, Estévez MC, Fabre N, Appeltans I, Martens T, Vandereyken K, Theobald H, Van Herck J, Petry P, Verheijden S, De Schepper S, Sifrim A, Liu Z, Ginhoux F, Azhar M, Schlitzer A, Matteoli G, Kierdorf K, Prinz M, Vanden Berghe P, Voet T, Boeckxstaens G. Dedicated macrophages organize and maintain the enteric nervous system. Nature 2023; 618:818-826. [PMID: 37316669 DOI: 10.1038/s41586-023-06200-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 05/11/2023] [Indexed: 06/16/2023]
Abstract
Correct development and maturation of the enteric nervous system (ENS) is critical for survival1. At birth, the ENS is immature and requires considerable refinement to exert its functions in adulthood2. Here we demonstrate that resident macrophages of the muscularis externa (MMϕ) refine the ENS early in life by pruning synapses and phagocytosing enteric neurons. Depletion of MMϕ before weaning disrupts this process and results in abnormal intestinal transit. After weaning, MMϕ continue to interact closely with the ENS and acquire a neurosupportive phenotype. The latter is instructed by transforming growth factor-β produced by the ENS; depletion of the ENS and disruption of transforming growth factor-β signalling result in a decrease in neuron-associated MMϕ associated with loss of enteric neurons and altered intestinal transit. These findings introduce a new reciprocal cell-cell communication responsible for maintenance of the ENS and indicate that the ENS, similarly to the brain, is shaped and maintained by a dedicated population of resident macrophages that adapts its phenotype and transcriptome to the timely needs of the ENS niche.
Collapse
Affiliation(s)
- Maria Francesca Viola
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marta Chavero-Pieres
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Elodie Modave
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Marcello Delfini
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Maria Cuende Estévez
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Naomi Fabre
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Tobie Martens
- Laboratory for Enteric NeuroScience, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Katy Vandereyken
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Hannah Theobald
- Quantitative Systems Biology, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Jens Van Herck
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Philippe Petry
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Simon Verheijden
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
- Janssen Research and Development, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sebastiaan De Schepper
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Alejandro Sifrim
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
- Laboratory of Multi-Omic Integrative Bioinformatics, Department of Genetics, KU Leuven, Leuven, Belgium
- Leuven AI Institute, KU Leuven, Leuven, Belgium
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Singapore Immunology Network, Agency for Science, Technology & Research, Singapore, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Mohamad Azhar
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Andreas Schlitzer
- Quantitative Systems Biology, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Laboratory for Intestinal Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.
- KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
48
|
Bernabè G, Shalata MEM, Zatta V, Bellato M, Porzionato A, Castagliuolo I, Brun P. Antibiotic Treatment Induces Long-Lasting Effects on Gut Microbiota and the Enteric Nervous System in Mice. Antibiotics (Basel) 2023; 12:1000. [PMID: 37370319 DOI: 10.3390/antibiotics12061000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The side effects of antibiotic treatment directly correlate with intestinal dysbiosis. However, a balanced gut microbiota supports the integrity of the enteric nervous system (ENS), which controls gastrointestinal neuromuscular functions. In this study, we investigated the long-term effects of antibiotic-induced microbial dysbiosis on the ENS and the impact of the spontaneous re-establishment of the gut microbiota on gastrointestinal functions. C57BL/6J mice were treated daily for two weeks with antibiotics. After 0-6 weeks of antibiotics wash-out, we determined (a) gut microbiota composition, (b) gastrointestinal motility, (c) integrity of the ENS, (d) neurochemical code, and (e) inflammation. Two weeks of antibiotic treatment significantly altered gut microbial composition; the genera Clostridium, Lachnoclostridium, and Akkermansia did not regain their relative abundance following six weeks of antibiotic discontinuation. Mice treated with antibiotics experienced delayed gastrointestinal transit and altered expression of neuronal markers. The anomalies of the ENS persisted for up to 4 weeks after the antibiotic interruption; the expression of neuronal HuC/D, glial-derived neurotrophic factor (Gdnf), and nerve growth factor (Ngf) mRNA transcripts did not recover. In this study, we strengthened the idea that antibiotic-induced gastrointestinal dysmotility directly correlates with gut dysbiosis as well as structural and functional damage to the ENS.
Collapse
Affiliation(s)
- Giulia Bernabè
- Department of Molecular Medicine, University of Padova, Via A. Gabelli, 63-35127 Padova, Italy
| | | | - Veronica Zatta
- Department of Molecular Medicine, University of Padova, Via A. Gabelli, 63-35127 Padova, Italy
| | - Massimo Bellato
- Department of Information Engineering, University of Padova, Via G. Gradenigo, 6-35131 Padova, Italy
| | - Andrea Porzionato
- Department of Neuroscience, University of Padova, Via A. Gabelli, 61-35127 Padova, Italy
| | - Ignazio Castagliuolo
- Department of Molecular Medicine, University of Padova, Via A. Gabelli, 63-35127 Padova, Italy
- Microbiology and Virology Unit of Padua University Hospital, School of Medicine, Via Ospedale, 1-35127 Padova, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padova, Via A. Gabelli, 63-35127 Padova, Italy
| |
Collapse
|
49
|
Mahapatra A, Dhakal A, Noguchi A, Vadlamani P, Hundley HA. ADARs employ a neural-specific mechanism to regulate PQM-1 expression and survival from hypoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539519. [PMID: 37205482 PMCID: PMC10187282 DOI: 10.1101/2023.05.05.539519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The ability to alter gene expression programs in response to changes in environmental conditions is central to the ability of an organism to thrive. For most organisms, the nervous system serves as the master regulator in communicating information about the animal's surroundings to other tissues. The information relay centers on signaling pathways that cue transcription factors in a given cell type to execute a specific gene expression program, but also provide a means to signal between tissues. The transcription factor PQM-1 is an important mediator of the insulin signaling pathway contributing to longevity and the stress response as well as impacting survival from hypoxia. Herein, we reveal a novel mechanism for regulating PQM-1 expression specifically in neural cells of larval animals. Our studies reveal that the RNA binding protein, ADR-1, binds to pqm-1 mRNA in neural cells. This binding is regulated by the presence of a second RNA binding protein, ADR-2, which when absent leads to reduced expression of both pqm-1 and downstream PQM-1 activated genes. Interestingly, we find that neural pqm-1 expression is sufficient to impact gene expression throughout the animal and affect survival from hypoxia; phenotypes that we also observe in adr mutant animals. Together, these studies reveal an important post-transcriptional gene regulatory mechanism that allows the nervous system to sense and respond to environmental conditions to promote organismal survival from hypoxia.
Collapse
Affiliation(s)
- Ananya Mahapatra
- Genome, Cell and Developmental Biology Graduate Program, Indiana University, Bloomington IN, 47405 USA
| | - Alfa Dhakal
- Cell, Molecular and Cancer Biology Graduate Program, Indiana University School of Medicine – Bloomington, Bloomington IN, 47405 USA
| | - Aika Noguchi
- Department of Biology, Indiana University, Bloomington IN 47405 USA
| | - Pranathi Vadlamani
- Medical Sciences Program, Indiana University School of Medicine – Bloomington, Bloomington IN, 47405 USA
| | | |
Collapse
|
50
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|